Tyrosine Kinase Inhibitors

Earn CME/CE in your profession:


Continuing Education Activity

Tyrosine kinase inhibitors (TKI) are a group of pharmacologic agents that disrupt the signal transduction pathways of protein kinases by several modes of inhibition. This activity will review the currently available drugs, their mechanism of action, routes of administration, indications, contraindications, and adverse effects.

Objectives:

  • Outline the different types of tyrosine kinase inhibitor (TKI) drugs and the currently available TKIs.
  • Describe the mechanism of action of tyrosine kinase inhibitors.
  • Summarize the indications and contraindications for each type of tyrosine kinase inhibitor.
  • Review the adverse effects and toxicity of tyrosine kinase inhibitors.

Indications

Mutations, dysregulation, and overexpression of protein kinases are involved in a multitude of disease processes. Around 1 in every 40 human genes codes for a protein kinase and nearly half of those genes map to either disease loci or cancer amplicons.[1] Interest in protein kinase inhibitors began with the FDA approval of the tyrosine kinase inhibitor (TKI) imatinib in 2001. Imatinib is an oral chemotherapy medication designed to target the BCR-Abl hybrid protein, a tyrosine kinase signaling protein produced in patients with Philadelphia-chromosome-positive chronic myelogenous leukemia. Since the introduction of Imatinib, the application of TKIs has been ever-expanding, particularly for cancer treatment, due to tyrosine kinases' critical roles in cellular signaling.[2][3]

Tyrosine kinase enzymes (TKs) can be categorized into receptor tyrosine kinases (RTKs), non-receptor tyrosine kinases (NRTKs), and a small group of dual-specificity kinases (DSK) which can phosphorylate serine, threonine, and tyrosine residues. RTKs are transmembrane receptor that includes vascular endothelial growth factor receptors (VEGFR), platelet-derived growth factor receptors (PDGFR), insulin receptor (InsR) family, and the ErbB receptor family, which includes epidermal growth factor receptors (EGFR) and the human epidermal growth factor receptor-2 (HER2). NRTKs are cytoplasmic proteins that consist of nine families, including Abl, Ack, Csk, Fak, Fes/Fer, Jak, Src, Syk/Zap70, and Tec, with the addition of Brl/Sik, Rak/Frk, Rlk/Txk, and Srm, which fall outside the nine defined families. The most notable example of DSKs is the mitogen-activated protein kinase kinases (MEKs), which are principally involved in the MAP pathways.[1][4][5]

As of now, there are over 50 FDA-approved TKIs. Comprehensive lists of FDA-approved TKIs with additional information are available at NIH PubChem and FDA.gov. Due to the broad reach of this topic and the rapid development of new drugs, this list is not fully comprehensive.

 

   TKI

Target

Indication

 

Deucravacitinib

TYK2

moderate-to-severe plaque psoriasis

 

Avapritinib         

PDGFR          

GIST (gastrointestinal stromal tumors) [6]

 

Capmatinib

c-MET

NSCLC (non-small cell lung cancer) [7]

 

Pemigatinib

FGFR

Cholangiocarcinoma with FGFR2 fusions or rearrangements [8]

 

Ripretinib

KIT/PDGFR

GIST [9]

 

Selpercatinib

RET

NSCLC and thyroid cancer [10]

 

Selumetinib

MEK1/2

Neurofibromatosis type 1 [11]

 

Tucatinib

Her2

HER2-positive breast cancer [12]

 

Entrectinib

TRKA/B/C, ROS1

ROS1-positive NSCLC, Solid tumors with NTRK fusion proteins [13]

 

Erdafitinib

FGFR

Urothelial bladder cancers [14]

 

Fedratinib

JAK2

Myelofibrosis [15]

 

Pexidartinib

CSF1R

Tenosynovial giant cell tumors [16]

 

Upadacitinib

PDGFR

GIST [17]

 

Zanubrutinib

BTK

Mantle cell lymphoma [18]

 

Baricitinib

JAK1/2

Rheumatoid arthritis [19]

 

Binimetinib

MEK1/2

Melanoma [20]

 

Dacomitinib

EGFR

EGFR-mutant NSCLC [21]

 

Fostamatinib

Syk

Chronic immune thrombocytopenia [22]

 

Gilteritinib

Flt

Acute myelogenous leukemia (AML) [23]

 

Larotrectinib

TRKA/B/C

Solid tumors with NTRK fusion proteins [24]

 

Lorlatinib

ALK

ALK-positive NSCLC [25]

 

Acalabrutinib

BTK

Mantle cell lymphoma, chronic lymphocytic  leukemia (CLL), small lymphocytic lymphomas [26]

 

Brigatinib

ALK

ALK-positive NSCLC [21]

 

Midostaurin

Flt3

AML, advanced systemic mastocytosis, mast cell leukemia [27]

 

Neratinib

HER2

HER2-positive breast cancer [21]

 

Alectinib

ALK, RET

ALK-positive NSCLC [28]

 

Cobimetinib

MEK1/2

combination therapy with vemurafenib for BRAF-positive melanoma [29]

 

Lenvatinib

VEGFR, RET

Differentiated thyroid cancer [30]

 

Osimertinib

EGFR

NSCLC [31]

 

Ceritinib

ALK

ALK-positive NSCLC resistant to crizotinib [32]

 

Nintedanib

FGFR

Idiopathic pulmonary fibrosis [33]

 

Afatinib

EGFR, HER2, ErB4

NSCLC [31]

 

Ibrutinib

BTK

CLL, graft vs. host disease, mantle cell lymphoma, marginal zone lymphoma [26]

 

Trametinib

MEK1/2

Melanoma [34]

 

Axitinib

VEGFR

Advanced renal cell carcinoma [35]

 

Bosutinib

BCR-Abl

Chronic myelogenous leukemia (CML) [36]

 

Cabozantinib

RET, VEGFR

Advanced medullary thyroid cancer, hepatocellular and renal cell carcinoma [37]

 

Ponatinib

BCR-Abl

Philadelphia chromosome-positive CML or acute lymphoblastic leukemia (ALL) [36]

 

Regorafenib

VEGFR

Colorectal cancer and hepatocellular carcinoma, GIST [37]

 

Tofacitinib

JAK3

Rheumatoid arthritis [19]

 

Crizotinib

ALK, ROS1

ALK or ROS1-positive NSCLC [37]

 

Ruxolitinib

JAK1/2/3, Tyk

Myelofibrosis, polycythemia vera [37]

 

Vandetanib

VEGFR2

Medullary thyroid cancer [37]

 

Pazopanib

VEGFR

Renal cell carcinoma, soft tissue sarcomas [37]

 

Lapatinib

EGFR, HER2

HER2-positive breast cancer [21]

 

Nilotinib

BCR-Abl

Philadelphia chromosome-positive CML [36]

 

Dasatinib

BCR-Abl

CML [36]

 

Sunitinib

VEGFR2

GIST, pancreatic neuroendocrine tumors, renal cell carcinomas [38]

 

Sorafenib

VEGFR

Hepatocellular carcinoma, renal cell carcinoma, differentiated thyroid cancer [3]

 

Erlotinib

EGFR

NSCLC, pancreatic cancer [31]

 

Gefitinib

EGFR

NSCLC [31]

 

Imatinib

BCR-Abl, KIT, PGFR

Philadelphia chromosome-positive CML or ALL, aggressive systemic mastocytosis, chronic eosinophilic leukemia, hypereosinophilic syndrome, dermatofibrosarcoma protuberans, GIST, myelodysplastic/myeloproliferative disease [36][39]

Mechanism of Action

As a whole, tyrosine kinases phosphorylate specific amino acids on substrate enzymes, which subsequently alter signal transduction leading to downstream changes in cellular biology. The downstream signal transduction set off by TKs can modify cell growth, migration, differentiation, apoptosis, and death. Constitutive activation or inhibition, either by mutations or other means, can lead to dysregulated signal cascades, potentially resulting in malignancy and other pathologies.[4][40] Therefore, blocking these initial signals via TKIs can prevent the aberrant action of the mutated or dysfunctional TKs.

Despite the diverse primary amino acid sequences, human kinases share similar 3D structures, particularly when it comes to the ATP-binding pocket located in the catalytically active region. The starting amino acid sequence (ASP-Phe-Gly or DFG) of the flexible activation loop that controls access to the activation site is also typically conserved.[41]

Kinase inhibitors are either irreversible or reversible. The irreversible kinase inhibitors tend to covalently bind and block the ATP site resulting in irreversible inhibition. The reversible kinase inhibitors can further subdivide into four major subtypes based on the confirmation of the binding pocket as well as the DFG motif.[3][42]

Below are listed various binding modes of TKIs.[3]

  • Type I inhibitors: competitively bind to the ATP-binding site of active TKs. The arrangement of the DFG motif in type I inhibitors has the aspartate residue facing the catalytic site of the kinase.
  • Type II inhibitors: bind to inactive kinases, usually at the ATP-binding site. The DFG motif in type II inhibitors protrudes outward away from the ATP-binding site. Due to the outward rotation of the DFG motif, many type II inhibitors can also exploit regions adjacent to the ATP-binding site that would otherwise be inaccessible.
  • Type III inhibitors: do not interact with the ATP-binding pocket. Type III inhibitors exclusively bind to allosteric pockets adjacent to the ATP-binding region.
  • Type IV inhibitors: bind allosteric sites far removed from the ATP-binding pocket.
  • Type V inhibitors: refer to a proposed subset of kinase inhibitors that exhibit multiple binding modes.[43]

Administration

Nearly all TKIs are effective when taken orally. Therapeutic loading and maintenance dosages are unique to each drug and should require unique dosing for each patient. When administering specific TKIs, many factors can contribute to reduced potency and the development of acquired resistance. These factors include such as whether or not food intake affects bioavailability, the mechanism of drug metabolism and elimination, liver and kidney function, drug-drug interactions, the presence of other medications that alter stomach pH, and patient demographics.[44][45][46]

Adverse Effects

Adverse events of TKIs are usually dose-based, with broad side effect profiles unique to each drug. However, due to similarities in drug targets, different classes of TKIs can have similar side effect profiles. Clinicians use BCR-Abl and KIT inhibitors to treat Philadelphia chromosome-positive CML and GIST, respectively.[47] Both KIT and BCR-Abl inhibitors, Imatinib, in particular, are known to cause adverse cutaneous drug reactions.[48]

EGFRs are a large family of RTKs associated with several cancers, including NSCL, breast, colorectal, pancreatic, esophageal, and head-and-neck cancers. The most common severe adverse effects associated with EGRF inhibitors are related to cutaneous adverse drug reactions.[48][49] The reason for this association is likely due to EGFRs’ role in normal skin integrity. Inhibition of EGFR hinders integumental function leading to dysfunctional epidermal differentiation and re-epithelialization, resulting in skin erosions.[49]

Angiogenesis is a critical step in cancer growth. VEGF is a key inducer of pathological angiogenesis expressed in nearly all human tumors.[50] Due to VEGF’s role in blood vessel survival and plasticity, TKIs that inhibit VEGFR carry associations with several cardiovascular toxicities, particularly hypertension.[51][52] This toxicity is likely because VEGF is necessary for adequate nitric oxide production. Inhibition of VEGF, therefore, results in elevated systemic vascular resistance.[53][54] Because VEGF is vital to endothelial cell survival, anti-VEGF therapy can diminish the integrity and regenerative capacity of endothelial cells, causing pro-coagulant changes. The long-term weakening and diminished integrity of blood vessel walls can eventually lead to thrombosis and hemorrhage.[55] Other adverse events associated with VEGFR TKIs include: renal injury, left ventricular dysfunction, cerebral and intestinal hemorrhage, cardiac ischemia, thrombosis, and skin reactions.[38]

Common adverse events related to TKIs are listed below. Not all adverse events are associated with every TKI, occurring at different frequencies depending on the drug.

 General

  • Fatigue
  • Fevers/Chills
  • Weight loss/gain

Gastrointestinal

  • Abdominal pain
  • Diarrhea
  • Dysgeusia
  • Constipation
  • Nausea/vomiting
  • Hepatotoxicity
  • Stomatitis

Cardiovascular

  • Hypertension
  • Hypotension
  • Congestive heart failure
  • Thrombosis
  • Cardiac ischemia
  • Myocardial infarction
  • Strokes
  • Intestinal hemorrhage
  • QT interval prolongation

Dermatological

  • Steven Johnson syndrome
  • Toxic epidermal necrolysis
  • Drug rash with eosinophilia and systemic symptoms
  • Acute generalized exanthematous pustulosis
  • Palmar-plantar erythrodysesthesia
  • Maculopapular rashes
  • Cheilitis
  • Facial edema
  • Eczema
  • Pruritis
  • Photosensitivity
  • Xerosis
  • Alopecia
  • Hair color changes
  • Paronychia

Endocrine

  • Hypokalemia
  • Thyroid dysfunction
  • Lacrimation

Hematologic

  • Anemia
  • Thrombocytopenia
  • Neutropenia
  • Bruising

Musculoskeletal

  • Myalgias
  • Arthralgia

Ophthalmic

  • Central serous retinopathy
  • Retinal pigment epithelial detachment
  • TKI keratitis

Renal

  • Kidney dysfunction
  • Proteinuria

Respiratory

  • Interstitial pulmonary disease
  • Pneumonia
  • Upper respiratory tract infection
  • Dyspnea
  • Epistaxis
  • Rhinorrhea

Neurological

  • Cognitive impairment
  • Peripheral neuropathy
  • Headaches
  • Dizziness

Contraindications

There are very few contraindications for TKIs. Considering the use of TKIs in life-extending cancer therapy, the benefits associated with TKI use generally outweigh the risks.[56] Data concerning TKI use in pregnancy is sparse; however, with rising rates of advanced maternal-age pregnancies, cancers requiring TKI therapy during pregnancy have become more common.[57][58] While there are several reports of successfully administering TKIs, such as erlotinib, imatinib, and nilotinib, during pregnancy, multiple studies demonstrate adverse events and teratogenic effects related to TKI use during pregnancy.[36][59][60][61]

Data on TKI use during pregnancy is still lacking in most cases. As a result, TKIs are typically co-prescribed with an effective contraception method during therapy and several weeks after discontinuing the TKI. Other limitations to using TKIs include severe adverse reactions. Patients at particular risk for one of the known adverse effects of the TKIs about to be prescribed, such as hypertension, interstitial lung disease, and long-QT syndrome, should receive an alternative therapy if possible.

Monitoring

While TKIs relieve the disease burden of most cancer patients, acquired resistance through various mechanisms remains a bottleneck in cancer targeted therapy. Patients require monitoring for disease progression after the initial benefit, which could be a sign of acquired resistance.[46] Genetic testing to identify known resistance mutations can also help guide genotype-directed therapy.

Toxicity

TKIs are generally well-tolerated, especially when compared to non-targeted cancer therapy. However, only a few of these drugs are selective for only one target. Due to the ubiquitous physiological role protein kinases play in the body, toxicities affecting various organs can occur. Organs commonly affected include the heart, lungs, liver, gastrointestinal tract, kidneys, thyroid, blood, and skin.[62]

The toxicity and efficacy of TKIs are often closely linked; this allows on-target toxic effects to act as biomarkers of effective pharmacological inhibition for certain TKIs. For example, skin rashes can serve as a monitoring mechanism for the effects of some TKIs that target EGFR and hypertension and can generally help monitor the inhibition of VEGFR.[63][64][65] However, the combined detrimental effects of both on-target and off-target toxicities can diminish a patient’s quality of life and limit the dose intensity of their medication, leading to sub-therapeutic treatment.

The optimal TKI of choice and dose is a requirement to reduce toxicities and adverse events. While TIKs are mainly administered at a fixed dose, several factors must guide dosing when devising an optimal TKI regimen. These factors include drug-drug/drug-food interactions, genetic polymorphisms of ABC transporters, patient adherence, intestinal absorption, distribution, metabolism, and elimination.[66][67] The interplay of multiple processes regulating the pharmacokinetics and pharmacodynamics of TKIs merits consideration when administering a TKI to titrate the optimal dosage.[68]

Enhancing Healthcare Team Outcomes

The development of TKI represents one of the most significant medical breakthroughs of the 21st century; however, one of the drawbacks of this drug class, endemic to small molecule therapies for cancer treatment in general, is the financial burden to the patient.[1] Kinase inhibitor therapy ranges from $5000 to $10,000 per month or more in the United States.[68] The significant financial burden may contribute to non-compliance resulting in disease progression and treatment resistance.[69] Clinicians, pharmacists, and other healthcare professionals should be aware of these financial burdens and discuss them along with the other potential physical toxicities of these drugs with the patient.

Given the non-specific nature of this drug class, it is imperative that therapy is ordered and subsequently followed through the collaborative efforts of an interprofessional team. This interprofessional team includes clinicians (MDs, DOs, NPs, PAs), specialists, nursing staff, and pharmacists. Each team member needs to be knowledgeable about the indications, dosing, and potential adverse effects of these drugs so they can counsel the patient regarding expectations and monitoring for adverse events, which will allow any needed changes in therapy to be made promptly, thereby driving optimal patient therapeutic outcomes. [Level 5]


Details

Updated:

7/18/2023 6:10:58 PM

References


[1]

Roskoski R Jr, Properties of FDA-approved small molecule protein kinase inhibitors: A 2020 update. Pharmacological research. 2020 Feb;     [PubMed PMID: 31862477]


[2]

Manning G,Whyte DB,Martinez R,Hunter T,Sudarsanam S, The protein kinase complement of the human genome. Science (New York, N.Y.). 2002 Dec 6;     [PubMed PMID: 12471243]


[3]

Wu P,Nielsen TE,Clausen MH, FDA-approved small-molecule kinase inhibitors. Trends in pharmacological sciences. 2015 Jul;     [PubMed PMID: 25975227]


[4]

Jiao Q,Bi L,Ren Y,Song S,Wang Q,Wang YS, Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Molecular cancer. 2018 Feb 19;     [PubMed PMID: 29455664]

Level 3 (low-level) evidence

[5]

Patterson KI,Brummer T,O'Brien PM,Daly RJ, Dual-specificity phosphatases: critical regulators with diverse cellular targets. The Biochemical journal. 2009 Mar 15;     [PubMed PMID: 19228121]


[6]

Gebreyohannes YK,Wozniak A,Zhai ME,Wellens J,Cornillie J,Vanleeuw U,Evans E,Gardino AK,Lengauer C,Debiec-Rychter M,Sciot R,Schöffski P, Robust Activity of Avapritinib, Potent and Highly Selective Inhibitor of Mutated KIT, in Patient-derived Xenograft Models of Gastrointestinal Stromal Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2019 Jan 15     [PubMed PMID: 30274985]


[7]

Vansteenkiste JF,Van De Kerkhove C,Wauters E,Van Mol P, Capmatinib for the treatment of non-small cell lung cancer. Expert review of anticancer therapy. 2019 Aug     [PubMed PMID: 31368815]


[8]

Liu PCC,Koblish H,Wu L,Bowman K,Diamond S,DiMatteo D,Zhang Y,Hansbury M,Rupar M,Wen X,Collier P,Feldman P,Klabe R,Burke KA,Soloviev M,Gardiner C,He X,Volgina A,Covington M,Ruggeri B,Wynn R,Burn TC,Scherle P,Yeleswaram S,Yao W,Huber R,Hollis G, INCB054828 (pemigatinib), a potent and selective inhibitor of fibroblast growth factor receptors 1, 2, and 3, displays activity against genetically defined tumor models. PloS one. 2020     [PubMed PMID: 32315352]


[9]

Activation of the alternative complement pathway due to resistance of zymosan-bound amplification convertase to endogenous regulatory mechanisms., Fearon DT,Austen KF,, Proceedings of the National Academy of Sciences of the United States of America, 1977 Apr     [PubMed PMID: 31205499]


[10]

Host specificity of a serum marker for hepatitis B: evidence that "e antigen" has the properties of an immunoglobulin., Neurath AR,Strick N,, Proceedings of the National Academy of Sciences of the United States of America, 1977 Apr     [PubMed PMID: 32557397]


[11]

Cell-free synthesis of the fourth component of guinea pig complement (C4): identification of a precursor of serum C4 (pro-C4)., Hall RE,Colten HR,, Proceedings of the National Academy of Sciences of the United States of America, 1977 Apr     [PubMed PMID: 28029918]


[12]

Lee A, Tucatinib: First Approval. Drugs. 2020 Jul     [PubMed PMID: 32548668]


[13]

Rolfo C,Ruiz R,Giovannetti E,Gil-Bazo I,Russo A,Passiglia F,Giallombardo M,Peeters M,Raez L, Entrectinib: a potent new TRK, ROS1, and ALK inhibitor. Expert opinion on investigational drugs. 2015     [PubMed PMID: 26457764]

Level 3 (low-level) evidence

[14]

Marandino L,Raggi D,Giannatempo P,Farè E,Necchi A, Erdafitinib for the treatment of urothelial cancer. Expert review of anticancer therapy. 2019 Oct     [PubMed PMID: 31544541]


[15]

Elimination of naturally occurring crosslinks in vaccinia virus DNA after viral penetration into cells., Pogo BG,, Proceedings of the National Academy of Sciences of the United States of America, 1977 Apr     [PubMed PMID: 31920387]


[16]

Voltage-dependent facilitation of Ca2+ entry in voltage-clamped, aequorin-injected molluscan neurons., Eckert R,Tillotson D,Ridgway EB,, Proceedings of the National Academy of Sciences of the United States of America, 1977 Apr     [PubMed PMID: 31602563]


[17]

Specific projection of displaced retinal ganglion cells upon the accessory optic system in the pigeon (Columbia livia)., Karten JH,Fite KV,Brecha N,, Proceedings of the National Academy of Sciences of the United States of America, 1977 Apr     [PubMed PMID: 31642025]


[18]

Syed YY, Zanubrutinib: First Approval. Drugs. 2020 Jan     [PubMed PMID: 31933167]


[19]

Reddy V,Cohen S, JAK Inhibitors: What Is New? Current rheumatology reports. 2020 Jul 22     [PubMed PMID: 32700001]


[20]

Queirolo P,Spagnolo F, Binimetinib for the treatment of NRAS-mutant melanoma. Expert review of anticancer therapy. 2017 Nov     [PubMed PMID: 28851243]


[21]

Turnover of 3H-5-hydroxytryptamine to 3H-5-hydroxyindoleacetic acid and the 3H-5-methoxyindoles in nondeprived and 24 hr food deprived rats., Kantak KM,Wayner MJ,Tilson HA,Sved A,, Pharmacology, biochemistry, and behavior, 1977 Feb     [PubMed PMID: 30649743]


[22]

The osteosarcomogenic effectiveness of the short-lived 224Ra compared with that of the long-lived 226Ra in mice., Müller WA,Luz A,, Radiation research, 1977 May     [PubMed PMID: 28967793]


[23]

[Incidence of septicemia in patients under prolonged mechanical respiration]., Domínguez de Villota E,Gracia Vidal MC,Barat Cascante G,Daza Pérez R,Muedra López A,Avello García F,, Revista clinica espanola, 1977 Mar 15     [PubMed PMID: 31528345]


[24]

Scott LJ, Larotrectinib: First Global Approval. Drugs. 2019 Feb     [PubMed PMID: 30635837]


[25]

Yang J,Gong W, Lorlatinib for the treatment of anaplastic lymphoma kinase-positive non-small cell lung cancer. Expert review of clinical pharmacology. 2019 Mar     [PubMed PMID: 30657349]


[26]

Liang C,Tian D,Ren X,Ding S,Jia M,Xin M,Thareja S, The development of Bruton's tyrosine kinase (BTK) inhibitors from 2012 to 2017: A mini-review. European journal of medicinal chemistry. 2018 May 10     [PubMed PMID: 29631132]


[27]

Effects of fatiguing isometric and isotonic exercise on fractionated patellar tendon reflex components., Morris AF,, Research quarterly, 1977 Mar     [PubMed PMID: 29487059]


[28]

Effects of a visually directed sensory-motor training program on depth perception of children., Newmeister GH,, Research quarterly, 1977 Mar     [PubMed PMID: 30030733]


[29]

Using gross motor activity to improve language arts concepts by third grade students., Penman KA,Christopher JR,Wood GS,, Research quarterly, 1977 Mar     [PubMed PMID: 27701080]


[30]

Cabanillas ME,Habra MA, Lenvatinib: Role in thyroid cancer and other solid tumors. Cancer treatment reviews. 2016 Jan     [PubMed PMID: 26678514]


[31]

Solassol I,Pinguet F,Quantin X, FDA- and EMA-Approved Tyrosine Kinase Inhibitors in Advanced {i}EGFR{/i}-Mutated Non-Small Cell Lung Cancer: Safety, Tolerability, Plasma Concentration Monitoring, and Management. Biomolecules. 2019 Oct 30     [PubMed PMID: 31671561]


[32]

Shaw AT,Engelman JA, Ceritinib in ALK-rearranged non-small-cell lung cancer. The New England journal of medicine. 2014 Jun 26     [PubMed PMID: 24963575]


[33]

Relationship of somatotype and body composition to physical performance in 7- to 12-year-old boys., Slaughter MH,Lohman TG,Misner JE,, Research quarterly, 1977 Mar     [PubMed PMID: 29327616]


[34]

Comparison of two nonparametric methods for estimating the reliability of motor performance tests., Stamm CL,Safrit MJ,, Research quarterly, 1977 Mar     [PubMed PMID: 30069762]


[35]

The effects of training and position on judges' ratings of a gymnastic event., Stephenson DA,Jackson AS,, Research quarterly, 1977 Mar     [PubMed PMID: 29033542]


[36]

Pallera A,Altman JK,Berman E,Abboud CN,Bhatnagar B,Curtin P,DeAngelo DJ,Gotlib J,Hagelstrom RT,Hobbs G,Jagasia M,Kantarjian HM,Kropf P,Metheny L,Moore JO,Ontiveros E,Purev E,Quiery A,Reddy VV,Rose MG,Shah NP,Smith BD,Snyder DS,Sweet KL,Tibes R,Yang DT,Gregory K,Sundar H,Deininger M,Radich JP, NCCN Guidelines Insights: Chronic Myeloid Leukemia, Version 1.2017. Journal of the National Comprehensive Cancer Network : JNCCN. 2016 Dec;     [PubMed PMID: 27956535]


[37]

Jeong W,Doroshow JH,Kummar S, United States Food and Drug Administration approved oral kinase inhibitors for the treatment of malignancies. Current problems in cancer. 2013 May-Jun     [PubMed PMID: 23972982]


[38]

Lankhorst S,Danser AH,van den Meiracker AH, Endothelin-1 and antiangiogenesis. American journal of physiology. Regulatory, integrative and comparative physiology. 2016 Feb 1;     [PubMed PMID: 26511523]

Level 2 (mid-level) evidence

[39]

Bhullar KS,Lagarón NO,McGowan EM,Parmar I,Jha A,Hubbard BP,Rupasinghe HPV, Kinase-targeted cancer therapies: progress, challenges and future directions. Molecular cancer. 2018 Feb 19     [PubMed PMID: 29455673]

Level 3 (low-level) evidence

[40]

Kolibaba KS,Druker BJ, Protein tyrosine kinases and cancer. Biochimica et biophysica acta. 1997 Dec 9;     [PubMed PMID: 9426205]


[41]

Tong M,Seeliger MA, Targeting conformational plasticity of protein kinases. ACS chemical biology. 2015 Jan 16;     [PubMed PMID: 25486330]


[42]

Norman RA,Toader D,Ferguson AD, Structural approaches to obtain kinase selectivity. Trends in pharmacological sciences. 2012 May;     [PubMed PMID: 22503441]


[43]

Lamba V,Ghosh I, New directions in targeting protein kinases: focusing upon true allosteric and bivalent inhibitors. Current pharmaceutical design. 2012;     [PubMed PMID: 22571662]


[44]

Wind S,Schnell D,Ebner T,Freiwald M,Stopfer P, Clinical Pharmacokinetics and Pharmacodynamics of Afatinib. Clinical pharmacokinetics. 2017 Mar;     [PubMed PMID: 27470518]


[45]

Kucharczuk CR,Ganetsky A,Vozniak JM, Drug-Drug Interactions, Safety, and Pharmacokinetics of EGFR Tyrosine Kinase Inhibitors for the Treatment of Non-Small Cell Lung Cancer. Journal of the advanced practitioner in oncology. 2018 Mar;     [PubMed PMID: 30588353]


[46]

Camidge DR,Pao W,Sequist LV, Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nature reviews. Clinical oncology. 2014 Aug;     [PubMed PMID: 24981256]


[47]

Jha P,Himanshu D,Jain N,Singh AK, Imatinib-induced Stevens-Johnsons syndrome. BMJ case reports. 2013 Jan 23;     [PubMed PMID: 23349042]

Level 3 (low-level) evidence

[48]

Ng CY,Chen CB,Wu MY,Wu J,Yang CH,Hui RC,Chang YC,Lu CW, Anticancer Drugs Induced Severe Adverse Cutaneous Drug Reactions: An Updated Review on the Risks Associated with Anticancer Targeted Therapy or Immunotherapies. Journal of immunology research. 2018;     [PubMed PMID: 29577050]


[49]

Chen CB,Wu MY,Ng CY,Lu CW,Wu J,Kao PH,Yang CK,Peng MT,Huang CY,Chang WC,Hui RC,Yang CH,Yang SF,Chung WH,Su SC, Severe cutaneous adverse reactions induced by targeted anticancer therapies and immunotherapies. Cancer management and research. 2018;     [PubMed PMID: 29844705]


[50]

Ferrara N, Vascular endothelial growth factor: basic science and clinical progress. Endocrine reviews. 2004 Aug;     [PubMed PMID: 15294883]


[51]

Carmeliet P,Ferreira V,Breier G,Pollefeyt S,Kieckens L,Gertsenstein M,Fahrig M,Vandenhoeck A,Harpal K,Eberhardt C,Declercq C,Pawling J,Moons L,Collen D,Risau W,Nagy A, Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996 Apr 4;     [PubMed PMID: 8602241]


[52]

Kamba T,McDonald DM, Mechanisms of adverse effects of anti-VEGF therapy for cancer. British journal of cancer. 2007 Jun 18;     [PubMed PMID: 17519900]


[53]

Escalante CP,Zalpour A, Vascular endothelial growth factor inhibitor-induced hypertension: basics for primary care providers. Cardiology research and practice. 2011;     [PubMed PMID: 21629798]


[54]

Horowitz JR,Rivard A,van der Zee R,Hariawala M,Sheriff DD,Esakof DD,Chaudhry GM,Symes JF,Isner JM, Vascular endothelial growth factor/vascular permeability factor produces nitric oxide-dependent hypotension. Evidence for a maintenance role in quiescent adult endothelium. Arteriosclerosis, thrombosis, and vascular biology. 1997 Nov;     [PubMed PMID: 9409257]


[55]

Kilickap S,Abali H,Celik I, Bevacizumab, bleeding, thrombosis, and warfarin. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2003 Sep 15;     [PubMed PMID: 12972536]


[56]

Rivas G,Llinás N,Bonilla C,Rubiano J,Cuello J,Arango N, Use of erlotinib throughout pregnancy: a case-report of a patient with metastatic lung adenocarcinoma. Lung cancer (Amsterdam, Netherlands). 2012 Aug;     [PubMed PMID: 22534670]

Level 3 (low-level) evidence

[57]

Dou X,Qin Y,Huang X,Jiang Q, Planned Pregnancy in Female Patients with Chronic Myeloid Leukemia Receiving Tyrosine Kinase Inhibitor Therapy. The oncologist. 2019 Nov;     [PubMed PMID: 31186377]

Level 2 (mid-level) evidence

[58]

Ji Y,Schwartz J,Hartford A,Ramsey J,Phillips J,Verschraegen C, Successful Treatment of Non-Small Cell Lung Cancer With Erlotinib Throughout Pregnancy. JAMA oncology. 2015 Sep;     [PubMed PMID: 26181671]


[59]

Berman E,Druker BJ,Burwick R, Chronic Myelogenous Leukemia: Pregnancy in the Era of Stopping Tyrosine Kinase Inhibitor Therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2018 Apr 20;     [PubMed PMID: 29447062]


[60]

Cortes JE,Abruzzese E,Chelysheva E,Guha M,Wallis N,Apperley JF, The impact of dasatinib on pregnancy outcomes. American journal of hematology. 2015 Dec;     [PubMed PMID: 26348106]


[61]

Pye SM,Cortes J,Ault P,Hatfield A,Kantarjian H,Pilot R,Rosti G,Apperley JF, The effects of imatinib on pregnancy outcome. Blood. 2008 Jun 15;     [PubMed PMID: 18322153]

Level 3 (low-level) evidence

[62]

Fujita KI,Ishida H,Kubota Y,Sasaki Y, Toxicities of Receptor Tyrosine Kinase Inhibitors in Cancer Pharmacotherapy: Management with Clinical Pharmacology. Current drug metabolism. 2017;     [PubMed PMID: 28059038]


[63]

Petrelli F,Borgonovo K,Cabiddu M,Lonati V,Barni S, Relationship between skin rash and outcome in non-small-cell lung cancer patients treated with anti-EGFR tyrosine kinase inhibitors: a literature-based meta-analysis of 24 trials. Lung cancer (Amsterdam, Netherlands). 2012 Oct;     [PubMed PMID: 22795701]

Level 1 (high-level) evidence

[64]

Estfan B,Byrne M,Kim R, Sorafenib in advanced hepatocellular carcinoma: hypertension as a potential surrogate marker for efficacy. American journal of clinical oncology. 2013 Aug;     [PubMed PMID: 22547010]


[65]

Hamnvik OP,Choueiri TK,Turchin A,McKay RR,Goyal L,Davis M,Kaymakcalan MD,Williams JS, Clinical risk factors for the development of hypertension in patients treated with inhibitors of the VEGF signaling pathway. Cancer. 2015 Jan 15;     [PubMed PMID: 25236375]


[66]

Klümpen HJ,Samer CF,Mathijssen RH,Schellens JH,Gurney H, Moving towards dose individualization of tyrosine kinase inhibitors. Cancer treatment reviews. 2011 Jun;     [PubMed PMID: 20833478]


[67]

Terada T,Noda S,Inui K, Management of dose variability and side effects for individualized cancer pharmacotherapy with tyrosine kinase inhibitors. Pharmacology     [PubMed PMID: 25976912]


[68]

Roskoski R Jr, Properties of FDA-approved small molecule protein kinase inhibitors. Pharmacological research. 2019 Jun;     [PubMed PMID: 30877063]


[69]

Dusetzina SB,Winn AN,Abel GA,Huskamp HA,Keating NL, Cost sharing and adherence to tyrosine kinase inhibitors for patients with chronic myeloid leukemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014 Feb 1;     [PubMed PMID: 24366936]